WO2003048197A1 - Production de virus, isolats viraux et vaccins - Google Patents

Production de virus, isolats viraux et vaccins Download PDF

Info

Publication number
WO2003048197A1
WO2003048197A1 PCT/NL2001/000892 NL0100892W WO03048197A1 WO 2003048197 A1 WO2003048197 A1 WO 2003048197A1 NL 0100892 W NL0100892 W NL 0100892W WO 03048197 A1 WO03048197 A1 WO 03048197A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
influenza
virus particle
alpha2
sialyltransferase
Prior art date
Application number
PCT/NL2001/000892
Other languages
English (en)
Inventor
Giuseppe Marzio
Maria Grazia Pau
Dirk Jan Elbertus Opstelten
Alphonsus Gerardus Cornelis Maria Uytdehaag
Original Assignee
Crucell Holland B.V.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Crucell Holland B.V. filed Critical Crucell Holland B.V.
Priority to PCT/NL2001/000892 priority Critical patent/WO2003048197A1/fr
Priority to AU2002225509A priority patent/AU2002225509A1/en
Priority to PCT/NL2002/000804 priority patent/WO2003048348A2/fr
Priority to KR1020047008520A priority patent/KR100979025B1/ko
Priority to EP02786227A priority patent/EP1465987B1/fr
Priority to ES02786227T priority patent/ES2297028T3/es
Priority to JP2003549527A priority patent/JP4480398B2/ja
Priority to AU2002351444A priority patent/AU2002351444B2/en
Priority to NZ533124A priority patent/NZ533124A/en
Priority to CA2468957A priority patent/CA2468957C/fr
Priority to AT02786227T priority patent/ATE384785T1/de
Priority to PT02786227T priority patent/PT1465987E/pt
Priority to DE60224843T priority patent/DE60224843T2/de
Priority to US10/497,832 priority patent/US7504248B2/en
Priority to CNB028243633A priority patent/CN100547069C/zh
Publication of WO2003048197A1 publication Critical patent/WO2003048197A1/fr
Priority to US11/102,073 priority patent/US7297680B2/en
Priority to US11/731,246 priority patent/US20070231860A1/en
Priority to US11/879,422 priority patent/US8802417B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/145Orthomyxoviridae, e.g. influenza virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0618Cells of the nervous system
    • C12N5/062Sensory transducers, e.g. photoreceptors; Sensory neurons, e.g. for hearing, taste, smell, pH, touch, temperature, pain
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • C12N2510/04Immortalised cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16151Methods of production or purification of viral material
    • C12N2760/16152Methods of production or purification of viral material relating to complementing cells and packaging systems for producing virus or viral particles
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16211Influenzavirus B, i.e. influenza B virus
    • C12N2760/16222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16211Influenzavirus B, i.e. influenza B virus
    • C12N2760/16234Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16211Influenzavirus B, i.e. influenza B virus
    • C12N2760/16251Methods of production or purification of viral material
    • C12N2760/16252Methods of production or purification of viral material relating to complementing cells and packaging systems for producing virus or viral particles

Definitions

  • the invention relates to the field of medicine.
  • the invention further relates to vaccines providing protection against influenza infection and to methods and means of obtaining these.
  • Influenza viruses are the etiological agents of flu, a highly contagious respiratory illness that has afflicted humans since ancient times. The virus was first identified in 1933, but numerous epidemics almost certainly attributable to influenza were reported throughout the centuries (Potter 1998). There have been three major cases of outbreaks of influenza in the last century. The so-called 'Spanish flu' of 1918 was particularly severe. It resulted in the death of an estimated 20 to 40 million people worldwide, the most severe recorded outbreak of human disease known in history. In 1957 the 'Asian flu' killed an estimated 1 million people, and in 1968 the 'Hong-Kong flu' was lethal for more than 700,000 individuals.
  • HA hemagglutinin
  • the HA gene was one of the first genes of the influenza virus to be identified and sequenced. It codes for a trans-membrane protein directly involved in attachment to and penetration into the host cell. HA initiates infection by binding to terminal sialyl-oligosaccharide receptor determinants present on glycoproteins and gangliosides present on the host cell surface. Terminal sialic acid residues of natural sialyl- glycoproteins and gangliosides are known to be the minimum determinants of binding. However, binding depends also on the type of sialic acid linkage to penultimate galactose and on the structure of more distant parts of the sialyl-glycoconjugate.
  • Human influenza viruses bind preferentially to receptors containing the sialic acid alpha-2,6-galactose (SAalpha2,6Gal) linkage, whereas avian viruses use the SAalpha2,3Gal linkage (reviewed in Suzuki 1994). This binding specificity determines also the cell tropism of the virus inside the host. Human influenza virus infection (and replication) are restricted to the respiratory tract, whereas avian influenza virus is found mainly in the cells lining the intestinal tract as well as in the lungs of birds.
  • SAalpha2,6Gal sialic acid alpha-2,6-galactose
  • SAalpha2,3Gal residues have a replicative advantage over virus variants that interact more specifically to SAalpha2,6Gal residues.
  • the SAalpha2,3Gal- specific virus variants are thus selected for in embryonated eggs (Gambaryan et al. 1999; Gambaryan et al. 1997).
  • Egg-adaptation not only increases the affinity for SAalpha2,3Gal, but it also results in decreased affinity for SAalpha2,6Gal.
  • HA in fact cannot accommodate both types of analogues equally well, and multiple mutations have been identified that confer this altered binding specificity (Daniels et al. 1987; Gambaryan et al. 1999; Ito et al. 1997; Suzuki et al. 1989).
  • Figure 1 Schematic representation of pAlpha2,6ST2000/Hygro.
  • Figure 2 Schematic representation of (A) pAlpha2,6STcDNA2000/Neo and (B) pAlpha2,6STcDNA2000/Hygro.
  • Figure 3 Schematic representation of (A) pAlpha2,3STcDNA2000/Neo and (B) pAlpha2,3STcDNA2000/Hygro.
  • Figure 5 Propagation of a primary clinical influenza isolate and a egg- passaged influenza batch (from the same primary isolate) on PER.C6 and PER.C6/alpha2,6ST, determined by fluorescence. Infectivity is expressed as percentage of cells positive for HA-immunofluorescent staining.
  • Figure 6 Propagation of a primary clinical influenza isolate and a egg- passaged influenza batch (from the same primary isolate) on PER.C6 and PER.C6/alpha2,6ST, determined by plaque assay. Infectivity is expressed as plaque-forming units (pfu's) per ml.
  • the present invention discloses methods for producing and/or propagating virus particles such as influenza virus particles that preferably are present in a virus isolate obtained from an infected subject, said method comprising the steps of: contacting a cell with a virus particle and culturing said cell under conditions conducive to propagation of said virus particle, wherein said cell over-expresses a nucleic acid encoding an alpha2,6 or an alpha2,3 sialyltransferase.
  • the invention also provides a method for selective propagation of a set of virus particles such as influenza virus particles present in an influenza isolate, wherein said set of virus particles has affinity for receptors comprising a specific glycosylation residue, said method comprising the steps of: incubating a cell with said isolate; culturing said cell under conditions conducive to propagation of said virus particle; and harvesting virus particles so produced from said cell and/or said culture medium.
  • a set of virus particles such as influenza virus particles present in an influenza isolate, wherein said set of virus particles has affinity for receptors comprising a specific glycosylation residue
  • the invention further provides novel vaccines and methods for making such vaccines, wherein said methods preferably comprise the steps of: treating the produced virus particles to yield antigenic parts; and harvesting at least one antigenic part such as hemagglutinin and/or neuraminidase from influenza virus.
  • the invention further provides cells and cell lines and the use thereof, that over-express certain proteins involved in glycosylation for the production of vaccines, e.g, vaccines against influenza infection.
  • Cells of the present invention are preferably human and transformed by adenovirus El, such as PER.C6 cells or derivatives thereof. DETAILED DESCRIPTION
  • the present invention provides methods for producing and/or propagating a virus particle, said method comprising the steps of: contacting a cell with a virus particle in a culture medium under conditions conducive to infection of said cell by said virus particle; and culturing said cell under conditions conducive to propagation of said virus particle, wherein said cell over-expresses a nucleic acid encoding an alpha2,6 sialyltransferase or a functional equivalent thereof.
  • Said nucleic acid may encode an alpha2,6 sialyltransferase from different sources, such as rat and human.
  • said alpha2,6 sialyltransferase is human alpha2,6 sialytransferase.
  • the invention further provides methods for producing and/or propagating a virus particle, said method comprising the steps of: contacting a cell with a virus particle in a culture medium under conditions conducive to infection of said cell by said virus particle; and culturing said cell under conditions conducive to propagation of said virus particle, wherein said cell over-expresses a nucleic acid encoding an alpha2,3 sialyltransferase or a functional equivalent thereof.
  • Said nucleic acid may encode an alpha2,3 sialyltransferase from different sources, such as rat and human.
  • said alpha2,3 sialyltransferase is human alpha2,3 sialytransferase.
  • said virus particle is an influenza virus particle.
  • the invention provides methods for propagating an influenza virus particle, wherein said influenza virus particle is present in an influenza isolate. More preferred are methods, wherein said influenza isolate is obtained from at least one influenza-infected mammahan subject. Even more preferred are methods for propagating an influenza virus particle, wherein said influenza-infected mammalian subject is human or pig. In another embodiment the invention provides methods for producing and/or propagating an influenza virus particle, wherein said influenza isolate is obtained from at least one influenza-infected bird. Isolates as used herein refers to batches of influenza viruses that are obtained from subjects that are infected with influenza viruses. These subjects may be all species that are susceptible for influenza viruses, such as humans, birds, pigs and horses.
  • Humans can get infected with influenza in different ways: either directly from other humans or directly from animal subjects such as pigs and birds.
  • Propagated viruses that are used for vaccine manufacturing might be originally derived from one or more subjects (one or more human individuals, or one or more birds, pigs, etc.).
  • influenza virus transmission from a bird to a human causes direct disease in humans, as was the case in the Hong Kong in 1997 (see above) it might be useful to be able to produce and/or propagate the influenza virus particles present in the bird isolate directly for vaccine manufacturing.
  • the present invention provides methods for producing and/or propagating influenza virus particles present in isolates that are obtained from species such as birds, pigs, horses and humans by over-expressing the sialyltransferase proteins that are involved in the glycosylation of cell surface proteins and that generate the so-called SAalpha2,3Gal and SAalpha2,6Gal linkages in the oligosaccharide chains.
  • Isolates as used herein preferably refers to clinical isolates (i.e., isolates obtained from diseased patients). Such clinical isolates are also referred to as primary isolates.
  • Primary isolates can be influenza isolates directly obtained from for instance the nose, mucus and/or faeces of humans or animals that are infected with influenza virus(es).
  • virus particles that are produced and/or propagated using the present invention may be present in passaged batches, but are preferably present in primary batches, such as clinical isolates.
  • the production and/or propagation of influenza virus particles is carried out by using cells in a culture medium, wherein said cell is transformed with El from adenovirus. More preferably, said cell is a human cell.
  • the invention provides methods for propagating an influenza virus particle according to the invention, wherein said human cell is PER.C6 or a derivative thereof.
  • PER.C6 cells are found to be useful for the propagation of different kinds of viruses such as rotavirus and influenza virus (see WO 01/38362).
  • PER.C6 cells were first generated by transforming cells obtained from an embryonal retina with the El region of Adenovirus serotype 5.
  • the present invention also provides methods for producing and/or propagating an influenza virus particle, wherein said nucleic acid encoding the sialyltransferase is heterologous to said cell.
  • said nucleic acid encoding the sialyltransferase is integrated into the genome of said cell.
  • Heterologous as used herein means that the nucleic acid is manipulated such that the gene encoding the sialyltransferase expresses more of the protein than without said manipulation.
  • Heterologous also means that the nucleic acid may be from a species that is different from the species from which the cell was derived, but may also be from the same species.
  • a cell is said to over-express the sialyltransferase when the cell expresses more sialyltransferase than typical for that cell.
  • a cell that over-expresses the sialyltransferase may also over-express the protein by manipulation of the genome of said cell such that the gene present in the genome of said cell expresses more of the protein than said cell did before it was manipulated.
  • the over-expression may be induced by external means such as integration of a different or more-active promoter, by removal or inhibition of suppressors that normally limit the expression of the protein, or by chemical means.
  • the over-expression may also be selected for. If cells are selected for a significant over-expression of at least one sialyltransferase they may be used for methods according to the present invention. Therefore, such cells and the use of such cells is also part of the present invention.
  • the present invention provides methods for making a vaccine, said method comprising the steps of: producing and/or propagating a virus particle according to methods of the invention; and inactivating the virus particles so produced.
  • said methods for making a vaccine further comprise the steps of: treating said virus particles so produced to yield antigenic parts; and obtaining at least one of said antigenic parts, preferably through means of purification and/or enrichment for said at least one part.
  • a purified and/or enriched composition comprising said at least one obtained antigenic part does not comprise other antigenic parts of said treated virus particles.
  • the invention provides methods for making a vaccine, wherein said antigenic part comprises the hemagglutinin protein or a part thereof, and/or the neuraminidase protein or a part thereof from influenza virus.
  • the neuraminidase (NA) and the hemagglutinin (HA) proteins are the most prominent antigenic parts of the influenza virus particle and are prone to differences during different propagation steps.
  • the invention also provides vaccines obtainable according to methods of the present invention, while it also provides pharmaceutical compositions comprising a vaccine obtainable according to the present invention.
  • the cells of the present invention are extremely useful for the propagation of primary, clinical isolates comprising influenza virus particles, while said cells can also be applied for propagating isolates that already have been passaged on embryonated eggs or on other systems, to obtain a selection of influenza virus particles that recognize specific glycosylation residues present on glycoproteins.
  • the present invention also provides the use of a cell line over-expressing an alpha2,6 sialyltransferase or a functional part thereof for the propagation of a virus particle and the use of a cell line over-expressing an alpha2,3 sialyltransferase or a functional part thereof for the propagation of a virus particle.
  • said virus particle is an influenza virus particle.
  • said influenza virus particle is present in an influenza isolate obtained from at least one influenza-infected mammalian subject. Even more preferred are uses of said cell line according to the present invention, wherein said influenza- infected mammalian subject is a human or a pig, whereas it is also preferred that said influenza virus particle is present in an influenza isolate obtained from at least one influenza-infected bird.
  • the present invention further provides a method for selective production and/or propagation of a set of predetermined virus particles present in an isolate, wherein said set of predetermined virus particles has a preference for a specific glycosylation moiety present on a receptor, and wherein said isolate comprises in addition to said set also virus particles not having said preference, said method comprising the steps of: incubating a cell which is capable of expressing and exposing said receptor comprising said specific glycosylation moiety, with said isolate in a culture medium under conditions conducive to infection of said cell by at least one virus particle present in said set; culturing said cell under conditions conducive to propagation of said virus particle; and harvesting virus particles so produced from said cell and/or said culture medium.
  • a glycosylation moiety as used herein refers to any kind of residue, linkage and or group of sugar types present in a oligosaccharide chain on a glycoprotein that is recognized by a virus particle for infection.
  • said glycosylation moiety comprises a SAalpha2,6Gal residue or a SAalpha2,3Gal residue.
  • said set of predetermined virus particles is a set of predetermined influenza virus particles.
  • the SAalpha2,6Gal residue and SAalpha2,3Gal residues are specifically recognized by the HA protein of the virus particle, in the case of influenza. It depends on the HA protein whether there is any specificity in the interaction with either one residue.
  • influenza isolates comprise viruses that interact specifically with the SAalpha2,6Gal residue as well as viruses that specifically interact with the SAalpha2,3Gal residue.
  • present invention it is now possible to selectively propagate either set of viruses from clinical, primary and/or passaged isolates to obtain propagated sets of viruses that are useful in the production of an influenza vaccine, useful in humans.
  • vaccines can be produced for humans
  • said influenza isolate is obtained from at least one influenza-infected human, pig or bird.
  • said cell is a human cell and that it is transformed with El from adenovirus.
  • Highly preferred are cells that are PER.C6 cells or derivatives thereof.
  • Derivatives as used herein refers to modified versions of the original PER.C6 cells, wherein for instance other heterologous nucleic acids are introduced, knocked-out, or in other ways modified.
  • Non-limiting examples of PER.C6 derivatives are PER.C6 cells that stable express a temperature sensitive mutant of
  • Adenovirus E2A or that express other adenovirus nucleic acids such as E4. If certain nucleic acids in PER.C6 cells have been switched on or off by other means such as chemical treatment or knock-out techniques, these cells still remain PER.C6 derivatives.
  • EPO erythropoietin
  • the invention provides methods for selective propagation of a set of virus particles present in an isolate, wherein said cell comprises a nucleic acid encoding a sialyltransferase that is heterologous to said cell.
  • said nucleic acid encoding a sialyltransferase is integrated into the genome of said cell.
  • Such an integrated nucleic acid is preferably stably integrated through the use of selection markers such as the hygromycin and neomycin resistance genes.
  • the present invention also provides human cells comprising a heterologous nucleic acid encoding an alpha2,6 sialyltransferase or an alpha2,3 sialyltransferase.
  • said nucleic acid is integrated into the genome of said human cell.
  • the invention also provides the use of such cells for the selective propagation of virus particles, preferably being influenza virus particles.
  • the present invention provides optimization of a process for propagation of primary isolates of human influenza virus. Also, the present invention provides optimization of a process for propagating primary as well as laboratory isolates of influenza viruses using the SAalpha2,6Gal or SAalpha2,3Gal (or both) glycosylation moieties present on cell surface glycoproteins.
  • human influenza viruses recognize the SAalpha2,6Gal moiety
  • the avian influenza viruses recognize the SAalpha2,3Gal moiety.
  • the swine influenza viruses generally utilize both residues.
  • the invention provides optimization of a process for propagation of any virus for which the replication depends on the activity of alpha2,3 sialyltransferase and/or alpha2,6 sialyltransferase, or more generally, on the presence of SAalpha2,3Gal or SAalpha2,6Gal residues.
  • the methods of the present invention comprise the use of cells in a culture medium. As an example of such a process, human cells were taken that are known to support efficient replication and production of influenza viruses.
  • the cells of the present invention are not only useful for the propagation of influenza viruses. It is well known in the art that other viruses such as Adeno-Associated Virus (AAV), human poliomavirus and parainfluenza viruses utilize the alpha2,3 and alpha2,6 linkages in glycoproteins for infection (Liu et al. 1998; Suzuki et al. 2001; Walters et al. 2001). Therefore the present invention also provides methods for (selective) production and/or propagation of other viruses that use these glycosylation structures for recognition and infection of the targeted cell. Furthermore, the invention provides the use of the cells of the invention and the methods and means for the production of viruses other than influenza and for the production of vaccines against such viruses, if applicable.
  • AAV Adeno-Associated Virus
  • human poliomavirus and parainfluenza viruses utilize the alpha2,3 and alpha2,6 linkages in glycoproteins for infection (Liu et al. 1998; Suzuki et al. 2001; Walters et al
  • the invention therefore also provides vaccines against viruses that utilize the SAalpha2,3Gal and the SAalpha2,6Gal residues for cellular recognition and infectivity. It has been previously demonstrated that PER.C6TM cells (ECACC deposit 96022940) represent an ideal substrate for the propagation of influenza virus and that the production levels from PER.C6 resulted in high-titer preparations suitable for vaccine purposes (WO 01/38362).
  • a novel cell line provided by the present invention named 'PER.C6-alpha2,6ST' is derived from PER.C6 through the following process: a plasmid harboring a nucleic acid encoding human alpha2,6 sialyltransferase under the control of the strong CMV promoter was transfected into PER.C6 cells and cells were subsequently selected for stable integration of the plasmid.
  • the PER.C6-alpha2,6ST cells are characterized by the higher expression of SAalpha2,6Gal-containing receptors as compared to the number of receptors carrying the SAalpha2,6Gal residue in the original PER.C6 cells.
  • PER.C6 cells are without over-expression of the alpha2,6 sialyltransferase already capable of expressing both SAalpha2,3Gal and SAalpha2,6Gal residues on cell surface glycoproteins. It is however an important aspect of the present invention to increase the percentage of proteins carrying the SAalpha2,6Gal residue in comparison to the percentage of proteins that carry the SAalpha2,3Gal residue ⁇ Due to direct substrate competition in the intracellular glycosylation machinery, receptors of the
  • SAalpha2,3Gal type become underrepresented on the cell surface of cells over- expressing the alpha2,6 sialyltransferase protein. These combined characteristics make this new cell line an ideal medium for propagating primary influenza virus isolates without inducing selection pressure in the wild type population.
  • the propagation of such isolates on the cells of the present invention results in efficient production of large virus stocks with unaltered HA specificity and immunogenicity that are highly useful for the production of vaccines.
  • virus produced in PER.C6-alpha2,6ST does not present mutations resulting from adaptation to the SAalpha2,3Gal receptor (as is the case for embryonated eggs) the immunogenic properties of this virus are most comparable with those of naturally circulating influenza viruses.
  • vaccine preparations obtained from influenza virus grown on PER.C6-alpha2,6ST are ideally suited to induce a protective response against circulating wild type influenza virus.
  • human influenza viruses are of the type recognizing the SAalpha2,6Gal linkages and it is therefore recognized in the art that it is desired to obtain vaccines comprising proteins from these viruses in order to sort a more protective immune response in humans (Newman et al. 1993).
  • PER.C6 cells have both SAalpha2,6Gal and SAalpha2,3Gal containing receptors at its surface. For a preferred propagation of the SAalpha2,6Gal recognizing viruses it is therefore preferred to have over-expression of receptors that harbor this component, as discussed above.
  • the present invention provides also methods and means for generating another novel cell line named 'PER.C6-alpha2,3ST'.
  • PER.C6-alpha2,3ST cells are derived from PER.C6 in a similar manner as described above for the PER.C6- alpha2,6ST cells, by transfection of a plasmid harboring nucleic acid encoding human alpha2,3 sialyltransferase under the control of the strong CMV promoter, after which cells carrying a stable integration of the plasmid are selected.
  • a PER.C6-alpha2,3ST cell is characterized by the higher expression of SAalpha2,3Gal-containing receptors.
  • alpha2,6 sialyltransferase and alpha2,3 sialyltransferase over- expressing cell lines are useful since alpha2,6 sialyltransferase over- expressing cells can be used for the propagation of influenza viruses that preferably recognize the SAalpha2,6Gal residue, while the alpha2,3 sialyltransferase over-expressing cells can be used for the propagation of influenza viruses that preferably recognize the SAalpha2,3Gal residue.
  • the infection and the spreading of the viruses mainly occurs via human-human contact and the viruses become more adapted to the infectious route via the SAalpha2,6Gal residues, then it is preferred to apply the alpha2,6 sialyltransferase over-expressing cell line.
  • alpha2,3 sialyltransferase or alpha2,3 sialyltransferase refer to the respective transferases and also to equivalents of said transferase, wherein said equivalents comprise the same transferase activity in kind not necessarily in amount as the transferase it is equivalent to.
  • Suitable equivalents can be generated by the person skilled in the art.
  • a part of said transferase is a suitable equivalent if it comprises the same transferase activity in kind not necessarily in amount.
  • Suitable equivalents are derivatives and/or analogues of alpha2,3 sialyltransferase or alpha2,3 sialyltransferase comprising the same transferase activity in kind not necessarily in amount as the transferase it is equivalent to. Such derivatives may be generated through conservative amino acid substitution or otherwise. A derivative can also be made from a part of the respective transferases.
  • An influenza virus particle as used herein can be an influenza virus or an influenza virus like particle.
  • An equivalent of an influenza virus particle is a virus(like) particle comprising the same infectivity properties in kind not necessarily in amount as an influenza virus particle. Such equivalents can for instance be generated by recombinant means. Such equivalents may comprise molecules not typically present in an influenza virus.
  • the fragment containing the sequence coding for alpha2,6 sialyltransferase was obtained by EcoRI digestion of plasmid pGST- Gal (a gift from Dr. I. van Die, Free University of Amsterdam;
  • the plasmid consists of a pBR322 backbone containing the entire cDNA sequence coding for rat alpha2,6 sialyltransferase, GenBank accession nr. M18769).
  • the fragment was made blunt-ended by T4 DNA polymerase according to standard procedures.
  • pcDNA2000/Hygro also known as plasmid pcDNA2000/Hyg(-) which has been described in WO 00/63403
  • pAlpha2,6ST2000/Hygro Figure 1
  • PER.C6-EPO were initially generated for other purposes, namely for experiments focusing on glycosylation of erythropoietin (EPO).
  • EPO is a protein involved in stimulation of erythropoiesis and its activity depends heavily on its sialic acid content for in vivo functionality.
  • the PER.C6-EPO cell line is a derivative of PER.C6 and overexpresses the human EPO protein (cells have been described in WO 00/63403). The fact that this cell line is producing EPO is not believed to be critical for the present invention.
  • PER.C6-EPO cells were cultured and transfected with pAlpha2,6ST2000/Hygro as described below.
  • PER.C6 cells were seeded in tissue culture dishes (10 cm diameter) with approximately 2-3 million cells/dish and were kept overnight at 37°C and 10% CO2. On the next day, cells are transfected using Lipofectamine (Gibco) according to the manufacturer's protocol. Twenty dishes were transfected each with 2 ⁇ g of pAlpha2,6ST2000/Hygro all according to standard protocols well known to persons skilled in the art. Another 6 dishes served as negative controls for hygromycin killing and transfection efficiency. On the next day, hygromycin was added to the dishes at a concentration of 50 ⁇ g/ml, dissolved in DMEM medium containing FBS.
  • Lipofectamine Gibco
  • the clones were subsequently tested for alpha2,6ST activity by FACS analysis on a FACsort apparatus (Becton Dickinson) using methods previously described by Govorkova et al. (1999).
  • FACS analysis on a FACsort apparatus Becton Dickinson
  • SAalpha2,6Gal-specific Sambucus nigra agglutinin DIG Glycan differentiation kit, Roche
  • DIG Glycan differentiation kit, Roche was used following the supplier's protocols.
  • These clones were subcultured in a time span of two months, during which FACS analysis experiments were performed on a regular basis to monitor expression of alpha2,6 sialyltransferase on the cell surface. Increased expression of SAalpha2,6Gal was stable.
  • a PCR fragment containing the full length cDNA of human alpha2,6 sialytransferase (GenBank accession nr: 14735135) is obtained by Polymerase Chain Reaction (PCR) on a human cDNA library using methods well known to persons skilled in the art.
  • the primers used for the amplification (sense: 5'- TTT TTT GGA TCC ATG ATT CAC ACC AAC CTG AAG AAA AAG-3', antisense: 5'-TTT TTT CTT AAG TTA GCA GTG AAT GGT CCG GAA GC-3') contain an additional 5' -tail that allows digestion with BamHI in the sense primer and Aflll in the antisense primer respectively.
  • PCR product is purified via agarose gel electrophoresis and digested with BamHI and Aflll and subsequently cloned into pcDNA2000/Hygro (described as pcDNA2000/Hyg(-) in WO 00/63403) and into pcDNA2000/Neo (this vector was basically constructed in the same way as pcDNA2000/Hyg(-) from pcDNA2000/DHFR as has been described in detail in WO 00/63403).
  • pcDNA2000/Hygro and pcDNA2000/Neo were also digested with BamHI and Aflll restriction enzymes.
  • the sequence and the correct cloning are checked by double stranded sequencing according to standard procedures known to persons skilled in the art of molecular biology.
  • the resulting plasmids are named pAlpha2,6STcDNA2000/Hygro ( Figure 2A) pAlpha2,6STcDNA2000/Neo ( Figure 2B). They comprise nucleic acid encoding human alpha2,6 sialyltransferase under the control of the extended CMV promoter (see WO 00/63403).
  • the plasmids confer resistance to neomycin and hygromycin respectively, that are used to select for clones that have integrated the plasmid into their genome in a stable manner.
  • the cDNA of human alpha2,3 sialyltransferase (GenBank accession nr. L23767) is obtained and cloned as described above for the human alpha2,6 sialyltransferase gene.
  • the primers that are used for the PCR reaction are: sense 5'-TTT TTT GGA TCC ATG TGT CCT GCA GGC TGG AAG CTC-3' and antisense 5'-TTT TTT CTT AAG TCA GAA GGA CGT GAG GTT CTT GAT AG-3'.
  • the resulting plasmids are named pAlpha2,3STcDNA2000/Hygro ( Figure 3A) pAlpha2,3STcDNA2000/Neo ( Figure 3B).
  • Example 4 Generation of stable PER.C6 cells over-expressing either human alpha2,6- or human alpha2,3 sialyltransferase.
  • Cells of the PER.C6 cell line are seeded in 40 tissue culture dishes (10 cm diameter) with approximately 2-3 million cells/dish and are kept overnight at 37°C and 10% CO2.
  • cells are transfected using Lipofectamine (Gibco) according to the manufacturer's protocol and to standard culturing procedures known to persons skilled in the art. Twenty dishes are transfected each with 5 ⁇ g of pAlpha2,6STcDNA2000/Neo. Another 20 dishes with non-transfected cells serve as negative controls for neomycin killing and transfection efficiency.
  • neomycin 0.5 mg/ml
  • medium containing FBS a medium containing FBS.
  • Cells are incubated over a period of 4-5 weeks, with regular washing of the cells with fresh medium supplemented with the selection agent. Cells are monitored daily for death, comparing with the negative controls that did not receive the plasmids harboring the neomycin and hygromycin selection markers. Outgrowing colonies are picked and subcultured generally as described for erythropoietin- and antibody-overexpressing cell lines in WO 00/63403. From each cell line, approximately 50 selected neomycin-resistent colonies are grown subsequently in 96-wells, 24-wells, 6-wells plates and T25 flask with neomycin.
  • each vial of each clone is frozen and stored for back-up.
  • Each clone is subsequently tested for production of recombinant human alpha2,6 sialyltransferase by FACS analysis using SAalpha2,6Gal-specific Sambucus nigra agglutinin as described above and as previously described by Govorkova et al. (1999).
  • the following selection of good producer clones is based on expression, culturing behaviour and viability. To allow checks for long term viability, suspension growth in roller bottles and bioreactor during extended time periods, more vials of the best performing clones are frozen, and are selected for further investigation.
  • Cell lines over-expressing the human alpha2,3 sialyltransferase protein are generated in generally the same way as described above for the human alpha2,6 sialyltransferase over-expressing PER.C6 cells.
  • plasmid pAlpha2,3STcDNA2000/Neo is used.
  • the resulting cell line is named PER.C6- H-alpha2,3 ST.
  • Example 5 Cell culture and infection with primary and adapted influenza virus isolates in PER.C6 cells and in alpha2,6 sialytransferase-overexpressing PER.C6 cells.
  • cells were seeded in 6-well plates, at the density of lxlO 6 cells/ml in a final volume of 2 ml of serum-free media, containing 2 mg/ml Pen/Strep (Gibco), 200 ng/ml Fungizone (Gibco) and 3 ⁇ g/ml trypsin-EDTA (Gibco).
  • Cells were infected with a viral inoculum of a primary isolate and with a PER.C6-adapted batch (derived from the primary isolate and passaged for 1 passage on PER.C6 cells).
  • the primary isolate that was used is the A/Netherlands/002/01 (H1N1, A/New Caledonia like, gift from Prof. Dr. A.
  • Example 6 Immunofluorescence test. Direct immunofluorescence (I.F.) assays for the detection of Influenza virus infection were carried out in infected cells (see above) using the IMAGENTM Influenza Virus A and B kit (Dako) according to the protocol provided by the supplier. Briefly, infected cells were centrifuged for 5 min. The supernatant was removed and the pellet resuspended in PBS. This was repeated twice to wash the cells thoroughly. The washed cell pellet was resuspended in PBS and 20 ⁇ l of cell suspension was added to each of two wells of an I.F. slide. This was allowed to dry at room temperature. The cells were fixed by adding 20 ⁇ l acetone to each well and air-dried..
  • I.F. Direct immunofluorescence
  • IMAGEN Influenza reagent i.e., labeled antibody specific Influenza A or B
  • the slide was then incubated for 15 min at 37°C on a damp tissue. Excess reagent was washed away with PBS and then rinsed for 5 min in PBS. The slide was air-dried at room temperature.
  • One drop of IMAGEN mounting fluid was added to each well and a cover slip placed over the slide (this was fixed in place with a small amount of nail polish). Samples were viewed microscopically using epifluorescence illumination. Infected cells were characterized by a bright apple-green fluorescence. The approximate percentage of cells that show positive (fluorescent green) compared with negative (red) cells was recorded. Results are shown in Figure 5. It is evident that PER.C6-alpha2,6 ST supported efficiently the replication of the clinical isolate (white bars).
  • Virus production in PER.C6 and PER.C6-alpha2,6 ST were studied by scoring for plaque formation in MDCK (Madin Darbin Canine Kidney) cells inoculated with virus supernatants. MDCK cells are particularly useful for such plaque assay experiments. A total of 1 ml of 10-fold diluted viral supernatants of primary and PER.C ⁇ -passaged influenza virus both propagated on PER.C6 and PER.C6-alpha2,6 ST according to the methods described in example 5, were inoculated on MDCK cells which were grown until 95% confluence in 6-well plates in DMEM supplemented with 2 mM L- glutamin.
  • the cells were washed twice with PBS and overloaded with 3 ml of agarose mix (1.2 ml 2.5% agarose, 1.5 ml 2x MEM, 30 ⁇ l 200 mM L-Glutamine, 24 ⁇ l trypsin-EDTA, 250 ⁇ l PBS). The cells were then incubated in a humid, 10% CO2 atmosphere at 37°C for approximately 3 days and viral plaques were visually scored and counted. Results are shown in

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Neurology (AREA)
  • General Engineering & Computer Science (AREA)
  • Pulmonology (AREA)
  • Molecular Biology (AREA)
  • Communicable Diseases (AREA)
  • Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Oncology (AREA)
  • Acoustics & Sound (AREA)
  • Analytical Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Neurosurgery (AREA)
  • Cell Biology (AREA)

Abstract

La présente invention porte sur des procédés de production et/ou de propagation sélective de particules virales telles que les particules virales de la grippe, prélevées de préférence sur un sujet infecté. Ce procédé consiste à mettre en contact et en culture une cellule qui surexprime alpha-2,6 ou alpha-2,3 sialyltransférase avec une particule virale dans des conditions conduisant à la propagation de cette particule virale. L'invention porte également sur l'utilisation de ces particules virales dans la préparation de vaccins.
PCT/NL2001/000892 1999-04-15 2001-12-07 Production de virus, isolats viraux et vaccins WO2003048197A1 (fr)

Priority Applications (18)

Application Number Priority Date Filing Date Title
PCT/NL2001/000892 WO2003048197A1 (fr) 2001-12-07 2001-12-07 Production de virus, isolats viraux et vaccins
AU2002225509A AU2002225509A1 (en) 2001-12-07 2001-12-07 Production and of viruses, viral isolates and vaccines
CA2468957A CA2468957C (fr) 2001-12-07 2002-12-09 Techniques visant la production de virus influenza dans des lignees cellulaires exprimant des sialyl transferases
PT02786227T PT1465987E (pt) 2001-12-07 2002-12-09 Produção de vírus, isolados virais e vacinas
EP02786227A EP1465987B1 (fr) 2001-12-07 2002-12-09 Production de virus, d'isolats viraux et de vaccins
ES02786227T ES2297028T3 (es) 2001-12-07 2002-12-09 Produccion de virus, aislados virales y vacunas.
JP2003549527A JP4480398B2 (ja) 2001-12-07 2002-12-09 ウイルス、ウイルス分離株及びワクチンの製造方法
AU2002351444A AU2002351444B2 (en) 2001-12-07 2002-12-09 Production of viruses, viral isolates and vaccines
NZ533124A NZ533124A (en) 2001-12-07 2002-12-09 Production of viruses, viral isolates and vaccines
PCT/NL2002/000804 WO2003048348A2 (fr) 2001-12-07 2002-12-09 Production de virus, d'isolats viraux et de vaccins
AT02786227T ATE384785T1 (de) 2001-12-07 2002-12-09 Herstellung von viren, virusisolaten, und impfstoffen
KR1020047008520A KR100979025B1 (ko) 2001-12-07 2002-12-09 바이러스, 바이러스 단리물 및 백신의 생산
DE60224843T DE60224843T2 (de) 2001-12-07 2002-12-09 Herstellung von viren, virusisolaten, und impfstoffen
US10/497,832 US7504248B2 (en) 2001-12-07 2002-12-09 Production of viruses viral isolates and vaccines
CNB028243633A CN100547069C (zh) 2001-12-07 2002-12-09 病毒、病毒分离物和疫苗的生产
US11/102,073 US7297680B2 (en) 1999-04-15 2005-04-08 Compositions of erythropoietin isoforms comprising Lewis-X structures and high sialic acid content
US11/731,246 US20070231860A1 (en) 1999-04-15 2007-03-30 Over-expression of enzymes involved in post-translational protein modifications in human cells
US11/879,422 US8802417B2 (en) 2001-12-07 2007-07-16 Production of viruses, viral isolates and vaccines

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/NL2001/000892 WO2003048197A1 (fr) 2001-12-07 2001-12-07 Production de virus, isolats viraux et vaccins

Publications (1)

Publication Number Publication Date
WO2003048197A1 true WO2003048197A1 (fr) 2003-06-12

Family

ID=19760783

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2001/000892 WO2003048197A1 (fr) 1999-04-15 2001-12-07 Production de virus, isolats viraux et vaccins

Country Status (2)

Country Link
AU (1) AU2002225509A1 (fr)
WO (1) WO2003048197A1 (fr)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003048348A2 (fr) * 2001-12-07 2003-06-12 Crucell Holland B.V. Production de virus, d'isolats viraux et de vaccins
WO2006070011A1 (fr) * 2004-12-30 2006-07-06 Crucell Holland B.V. Procedes d'obtention de proteines recombinantes a sialylation accrue a partir de cellules qui expriment une proteine adenovirale e1a et proteines obtenues de cette maniere
US7132280B2 (en) 1999-04-15 2006-11-07 Crucell Holland, B.V. Recombinant protein production in a human cell
US7192759B1 (en) 1999-11-26 2007-03-20 Crucell Holland B.V. Production of vaccines
WO2007045674A1 (fr) * 2005-10-21 2007-04-26 Crucell Holland B.V. Vaccins de production du virus de la grippe
US7291484B2 (en) 2003-05-09 2007-11-06 Crucell Holland B.V. Processes for culturing E1-immortalized cells to increase product yields
US7527961B2 (en) 1999-11-26 2009-05-05 Crucell Holland B.V. Production of vaccines
US7604960B2 (en) 1999-04-15 2009-10-20 Crucell Holland B.V. Transient protein expression methods
US7642078B2 (en) 2005-12-28 2010-01-05 Crucell Holland B.V. Methods to obtain recombinant proteins with increased sialylation from cells that express adenovirus E1A protein, and proteins obtained thereby
US8236293B2 (en) 1995-06-15 2012-08-07 Crucell Holland B.V. Means and methods for nucleic acid delivery vehicle design and nucleic acid transfer
US8236561B2 (en) 1999-04-15 2012-08-07 Crucell Holland B.V. Efficient production of IgA in recombinant mammalian cells
AU2011203144B2 (en) * 2004-12-30 2012-08-16 Crucell Holland B.V. Methods to obtain recombinant proteins with increased sialylation from cells that express adenovirus E1A protein, and proteins obtained thereby
US8524477B2 (en) 2002-10-29 2013-09-03 Crucell Holland B.V. Methods to obtain recombinant proteins with increased sialylation from cells that express adenovirus E1A protein, and proteins obtained thereby

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000063403A2 (fr) * 1999-04-15 2000-10-26 Crucell Holland B.V. Production de proteine de recombinaison dans une cellule humaine
WO2001038362A2 (fr) * 1999-11-26 2001-05-31 Crucell Holland B.V. Fabrication de vaccins

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000063403A2 (fr) * 1999-04-15 2000-10-26 Crucell Holland B.V. Production de proteine de recombinaison dans une cellule humaine
WO2001038362A2 (fr) * 1999-11-26 2001-05-31 Crucell Holland B.V. Fabrication de vaccins

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CARROLL S M ET AL: "DIFFERENTIAL INFECTION OF RECEPTOR-MODIFIED HOST CELLS BY RECEPTOR-SPECIFIC INFLUENZA VIRUSES", VIRUS RESEARCH, vol. 3, no. 2, 1985, pages 165 - 180, XP001095308, ISSN: 0168-1702 *
PAU M G ET AL: "The human cell line PER.C6 provides a new manufacturing system for the production of influenza vaccines", VACCINE, BUTTERWORTH SCIENTIFIC. GUILDFORD, GB, vol. 19, no. 17-19, 2001, pages 2716 - 2721, XP002201217, ISSN: 0264-410X *

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8236293B2 (en) 1995-06-15 2012-08-07 Crucell Holland B.V. Means and methods for nucleic acid delivery vehicle design and nucleic acid transfer
US7604960B2 (en) 1999-04-15 2009-10-20 Crucell Holland B.V. Transient protein expression methods
US7132280B2 (en) 1999-04-15 2006-11-07 Crucell Holland, B.V. Recombinant protein production in a human cell
US8236561B2 (en) 1999-04-15 2012-08-07 Crucell Holland B.V. Efficient production of IgA in recombinant mammalian cells
US7833753B2 (en) 1999-04-15 2010-11-16 Crucell Holland B.V. Methods of producing erythropoietin isoforms comprising Lewis-X structures and high sialic acid content and compositions of the same
US7297680B2 (en) 1999-04-15 2007-11-20 Crucell Holland B.V. Compositions of erythropoietin isoforms comprising Lewis-X structures and high sialic acid content
US7470523B2 (en) 1999-04-15 2008-12-30 Crucell Holland B.V. Recombinant protein production in a human cell
US7491532B2 (en) 1999-04-15 2009-02-17 Crucell Holland, B.V. Recombinant protein production in a human cell
US7192759B1 (en) 1999-11-26 2007-03-20 Crucell Holland B.V. Production of vaccines
US7964198B2 (en) 1999-11-26 2011-06-21 Crucell Holland B.V. Production of vaccines
US7527961B2 (en) 1999-11-26 2009-05-05 Crucell Holland B.V. Production of vaccines
US7521220B2 (en) 1999-11-26 2009-04-21 Crucell Holland B.V. Production of vaccines
US7833788B2 (en) 1999-11-26 2010-11-16 Crucell Holland B.V. Production of vaccines
US7550284B2 (en) 1999-11-26 2009-06-23 Crucell Holland B.V. Production of vaccines
WO2003048348A3 (fr) * 2001-12-07 2003-12-11 Crucell Holland Bv Production de virus, d'isolats viraux et de vaccins
WO2003048348A2 (fr) * 2001-12-07 2003-06-12 Crucell Holland B.V. Production de virus, d'isolats viraux et de vaccins
US8802417B2 (en) 2001-12-07 2014-08-12 Crucell Holland B.V. Production of viruses, viral isolates and vaccines
US7504248B2 (en) 2001-12-07 2009-03-17 Crucell Holland B.V. Production of viruses viral isolates and vaccines
US8524477B2 (en) 2002-10-29 2013-09-03 Crucell Holland B.V. Methods to obtain recombinant proteins with increased sialylation from cells that express adenovirus E1A protein, and proteins obtained thereby
US8008043B2 (en) 2003-05-09 2011-08-30 Crucell Holland B.V. Cultures of E1-immortalized cells and processes for culturing the same to increase product yields therefrom
US7608431B2 (en) 2003-05-09 2009-10-27 Crucell Holland B.V. Fed-batch process for production of erythropoietin in human embryonic retina cells that express adenovirus E1A
US7291484B2 (en) 2003-05-09 2007-11-06 Crucell Holland B.V. Processes for culturing E1-immortalized cells to increase product yields
AU2005321246B2 (en) * 2004-12-30 2011-04-14 Crucell Holland B.V. Methods to obtain recombinant proteins with increased sialylation from cells that express adenovirus E1A protein, and proteins obtained thereby
EP2402450A1 (fr) * 2004-12-30 2012-01-04 Crucell Holland B.V. Procédés pour obtenir des protéines recombinantes dotées d'une structure LacdiNac diminuée
WO2006070011A1 (fr) * 2004-12-30 2006-07-06 Crucell Holland B.V. Procedes d'obtention de proteines recombinantes a sialylation accrue a partir de cellules qui expriment une proteine adenovirale e1a et proteines obtenues de cette maniere
AU2011203144B2 (en) * 2004-12-30 2012-08-16 Crucell Holland B.V. Methods to obtain recombinant proteins with increased sialylation from cells that express adenovirus E1A protein, and proteins obtained thereby
CN102653772A (zh) * 2004-12-30 2012-09-05 克鲁塞尔荷兰公司 从表达腺病毒e1a蛋白的细胞中获得唾液酸化增加的重组蛋白质的方法及由此获得的蛋白质
KR101319733B1 (ko) 2004-12-30 2013-10-23 크루셀 홀란드 비.브이. 아데노바이러스 e1a 단백질을 발현하는 세포로부터시알릴화가 증가된 재조합 단백질의 제조 방법, 및 그로인해 얻어진 단백질들
WO2007045674A1 (fr) * 2005-10-21 2007-04-26 Crucell Holland B.V. Vaccins de production du virus de la grippe
US7642078B2 (en) 2005-12-28 2010-01-05 Crucell Holland B.V. Methods to obtain recombinant proteins with increased sialylation from cells that express adenovirus E1A protein, and proteins obtained thereby

Also Published As

Publication number Publication date
AU2002225509A1 (en) 2003-06-17

Similar Documents

Publication Publication Date Title
US8802417B2 (en) Production of viruses, viral isolates and vaccines
US7964198B2 (en) Production of vaccines
JP6352974B2 (ja) ワクチンのための高力価組み換えインフルエンザ・ウィルス
JP4331432B2 (ja) ワクチンの生成方法
US7527961B2 (en) Production of vaccines
EP2233152A1 (fr) Virus de la grippe A réassorti et de croissance haute
WO2003048197A1 (fr) Production de virus, isolats viraux et vaccins
US20140377308A1 (en) METHOD FOR PRODUCTION OF pH STABLE ENVELOPED VIRUSES
Marzio et al. Production of viruses, viral isolates and vaccines
WO2010046335A1 (fr) Production du virus influenza par génétique inverse dans les cellules per.c6 en conditions asériques

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP